Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Mol Pharm ; 18(12): 4428-4436, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34649437

RESUMEN

Type 1 diabetes develops in childhood and adolescence, with peak incidence in the early teenage years. There is an urgent need for an accurate method to detect insulin-producing ß-cells in patients that is not affected by alterations in ß-cell function. As part of our research program to design specific probes to measure ß-cell mass, we recently developed a novel insulin-binding peptide probe (IBPP) for the detection of ß-cells in vivo. Here, we applied our innovative method to show specific labeling of this IBPP to human and mouse fixed ß-cells in pancreatic islets. Importantly, we showed staining of human and mouse islets in culture without any negative functional or cell viability impact. Moreover, the IBPP-stained mouse islets after tail vein injection in vivo, albeit with batch differences in staining efficiency. In conclusion, we provide evidence showing that the IBPP can be used for future accurate detection of ß-cell mass in a variety of preclinical models of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/diagnóstico por imagen , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Células Cultivadas , Humanos , Insulina/análisis , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Coloración y Etiquetado
2.
Biomacromolecules ; 18(12): 4249-4260, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29035554

RESUMEN

Protein aggregation into amyloid fibrils is a ubiquitous phenomenon across the spectrum of neurodegenerative disorders and type 2 diabetes. A common strategy against amyloidogenesis is to minimize the populations of toxic oligomers and protofibrils by inhibiting protein aggregation with small molecules or nanoparticles. However, melanin synthesis in nature is realized by accelerated protein fibrillation to circumvent accumulation of toxic intermediates. Accordingly, we designed and demonstrated the use of star-shaped poly(2-hydroxyethyl acrylate) (PHEA) nanostructures for promoting aggregation while ameliorating the toxicity of human islet amyloid polypeptide (IAPP), the peptide involved in glycemic control and the pathology of type 2 diabetes. The binding of PHEA elevated the ß-sheet content in IAPP aggregates while rendering a new morphology of "stelliform" amyloids originating from the polymers. Atomistic molecular dynamics simulations revealed that the PHEA arms served as rodlike scaffolds for IAPP binding and subsequently accelerated IAPP aggregation by increased local peptide concentration. The tertiary structure of the star nanoparticles was found to be essential for driving the specific interactions required to impel the accelerated IAPP aggregation. This study sheds new light on the structure-toxicity relationship of IAPP and points to the potential of exploiting star polymers as a new class of therapeutic agents against amyloidogenesis.


Asunto(s)
Amiloide/química , Proteínas Amiloidogénicas/química , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polímeros/química , Agregación Patológica de Proteínas/patología , Amiloidosis/patología , Animales , Línea Celular , Diabetes Mellitus Tipo 2/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación de Dinámica Molecular , Nanopartículas/química
3.
Small ; 12(12): 1615-26, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-26808649

RESUMEN

Human islet amyloid polypeptide (hIAPP, or amylin) forms amyloid deposits in the islets of Langerhans, a phenomenon that is associated with type-2 diabetes impacting millions of people worldwide. Accordingly, strategies against hIAPP aggregation are essential for the prevention and eventual treatment of the disease. Here, it is shown that generation-3 OH-terminated poly(amidoamine) dendrimer, a polymeric nanoparticle, can effectively halt the aggregation of hIAPP and shut down hIAPP toxicity in pancreatic MIN6 and NIT-1 cells as well as in mouse islets. This finding is supported by high-throughput dynamic light scattering experiment and thioflavin T assay, where the rapid evolution of hIAPP nucleation and elongation processes is halted by the addition of the dendrimer up to 8 h. Discrete molecular dynamics simulations further reveal that hIAPP residues bound strongly with the dendrimer near the c-terminal portion of the peptide, where the amyloidogenic sequence (residues 22-29) locates. Furthermore, simulations of hIAPP dimerization reveal that binding with the dendrimer significantly reduces formation of interpeptide contacts and hydrogen bonds, thereby prohibiting peptide self-association and amyloidosis. This study points to a promising nanomedicinal strategy for combating type-2 diabetes and may have broader implications for targeting neurological disorders whose distinct hallmark is also amyloid fibrillation.


Asunto(s)
Amiloide/metabolismo , Dendrímeros/toxicidad , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Agregado de Proteínas/efectos de los fármacos , Benzotiazoles , Muerte Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Humanos , Hidroxilación , Células Secretoras de Insulina/efectos de los fármacos , Modelos Moleculares , Multimerización de Proteína/efectos de los fármacos , Tiazoles/metabolismo
4.
Phys Chem Chem Phys ; 18(1): 94-100, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26625841

RESUMEN

Human islet amyloid polypeptide (hIAPP or amylin) aggregation is directly associated with pancreatic ß-cell death and subsequent insulin deficiency in type 2 diabetes (T2D). Since no cure is currently available for T2D, it is of great benefit to devise new anti-aggregation molecules, which protect ß-cells against hIAPP aggregation-induced toxicity. Engineered nanoparticles have been recently exploited as anti-aggregation nanomedicines. In this work, we studied graphene oxide (GO) nanosheets for their potential for hIAPP aggregation inhibition by combining computational modeling, biophysical characterization and cell toxicity measurements. Using discrete molecular dynamics (DMD) simulations and in vitro studies, we showed that GO exhibited an inhibitory effect on hIAPP aggregation. DMD simulations indicated that the strong binding of hIAPP to GO nanosheets was driven by hydrogen bonding and aromatic stacking and that the strong peptide-GO binding efficiently inhibited hIAPP self-association and aggregation on the nanosheet surface. Secondary structural changes of hIAPP upon GO binding derived from DMD simulations were consistent with circular dichroism (CD) spectroscopy measurements. Transmission electron microscopy (TEM) images confirmed the reduction of hIAPP aggregation in the presence of GO. Furthermore, we carried out a cell toxicity assay and found that these nanosheets protected insulin-secreting NIT-1 pancreatic ß-cells against hIAPP-induced toxicity. Our multidisciplinary study suggests that GO nanosheets have the potential to be utilized as an anti-aggregation nanomedicine itself in addition to a biosensor or delivery vehicle for the mitigation of T2D progression.


Asunto(s)
Grafito/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/biosíntesis , Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Óxidos/farmacología , Agregado de Proteínas/efectos de los fármacos , Línea Celular , Grafito/química , Humanos , Células Secretoras de Insulina/metabolismo , Simulación de Dinámica Molecular , Óxidos/química
5.
J Mol Endocrinol ; 59(4): 325-337, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28827413

RESUMEN

Type 1 diabetes (T1D) is characterized by the destruction of insulin-producing ß-cells by immune cells in the pancreas. Pro-inflammatory including TNF-α, IFN-γ and IL-1ß are released in the islet during the autoimmune assault and signal in ß-cells through phosphorylation cascades, resulting in pro-apoptotic gene expression and eventually ß-cell death. Protein tyrosine phosphatases (PTPs) are a family of enzymes that regulate phosphorylative signalling and are associated with the development of T1D. Here, we observed expression of PTPN6 and PTPN1 in human islets and islets from non-obese diabetic (NOD) mice. To clarify the role of these PTPs in ß-cells/islets, we took advantage of CRISPR/Cas9 technology and pharmacological approaches to inactivate both proteins. We identify PTPN6 as a negative regulator of TNF-α-induced ß-cell death, through JNK-dependent BCL-2 protein degradation. In contrast, PTPN1 acts as a positive regulator of IFN-γ-induced STAT1-dependent gene expression, which enhanced autoimmune destruction of ß-cells. Importantly, PTPN1 inactivation by pharmacological modulation protects ß-cells and primary mouse islets from cytokine-mediated cell death. Thus, our data point to a non-redundant effect of PTP regulation of cytokine signalling in ß-cells in autoimmune diabetes.


Asunto(s)
Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal , Animales , Muerte Celular/genética , Muerte Celular/inmunología , Expresión Génica , Técnicas de Inactivación de Genes , Marcación de Gen , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos NOD , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Fosfatasas/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Diabetes ; 66(12): 2973-2986, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28928277

RESUMEN

The members of the BCL-2 family are crucial regulators of the mitochondrial pathway of apoptosis in normal physiology and disease. Besides their role in cell death, BCL-2 proteins have been implicated in the regulation of mitochondrial oxidative phosphorylation and cellular metabolism. It remains unclear, however, whether these proteins have a physiological role in glucose homeostasis and metabolism in vivo. In this study, we report that fat accumulation in the liver increases c-Jun N-terminal kinase-dependent BCL-2 interacting mediator of cell death (BIM) expression in hepatocytes. To determine the consequences of hepatic BIM deficiency in diet-induced obesity, we generated liver-specific BIM-knockout (BLKO) mice. BLKO mice had lower hepatic lipid content, increased insulin signaling, and improved global glucose metabolism. Consistent with these findings, lipogenic and lipid uptake genes were downregulated and lipid oxidation enhanced in obese BLKO mice. Mechanistically, BIM deficiency improved mitochondrial function and decreased oxidative stress and oxidation of protein tyrosine phosphatases, and ameliorated activation of peroxisome proliferator-activated receptor γ/sterol regulatory element-binding protein 1/CD36 in hepatocytes from high fat-fed mice. Importantly, short-term knockdown of BIM rescued obese mice from insulin resistance, evidenced by reduced fat accumulation and improved insulin sensitivity. Our data indicate that BIM is an important regulator of liver dysfunction in obesity and a novel therapeutic target for restoring hepatocyte function.


Asunto(s)
Proteína 11 Similar a Bcl2/fisiología , Hígado Graso/etiología , Resistencia a la Insulina , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Hígado/metabolismo , Obesidad/metabolismo , Estrés Oxidativo , Animales , Células Cultivadas , Activación Enzimática , Ácidos Grasos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
7.
FEBS J ; 283(16): 3002-15, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26972840

RESUMEN

Diabetes mellitus are complex, multi-organ metabolic pathologies characterized by hyperglycemia. Emerging evidence shows that the highly conserved and potent JAK/STAT signaling pathway is required for normal homeostasis, and, when dysregulated, contributes to the development of obesity and diabetes. In this review, we analyze the role of JAK/STAT activation in the brain, liver, muscle, fat and pancreas, and how this affects the course of the disease. We also consider the therapeutic implications of targeting the JAK/STAT pathway in treatment of obesity and diabetes.


Asunto(s)
Diabetes Mellitus/metabolismo , Quinasas Janus/metabolismo , Obesidad/metabolismo , Factores de Transcripción STAT/metabolismo , Tejido Adiposo/enzimología , Tejido Adiposo/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Diabetes Mellitus/enzimología , Diabetes Mellitus/terapia , Hígado Graso/metabolismo , Humanos , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/metabolismo , Quinasas Janus/antagonistas & inhibidores , Ratones , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Obesidad/enzimología , Obesidad/terapia , Factores de Transcripción STAT/antagonistas & inhibidores , Transducción de Señal
8.
Sci Rep ; 6: 23802, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27033313

RESUMEN

BCL-2 proteins have been implicated in the control of glucose homeostasis and metabolism in different cell types. Thus, the aim of this study was to determine the role of the pro-apoptotic BH3-only protein, p53-upregulated-modulator-of-apoptosis (PUMA), in metabolic changes mediated by diet-induced obesity, using PUMA deficient mice. At 10 weeks of age, knockout and wild type mice either continued consuming a low fat chow diet (6% fat), or were fed with a high fat diet (23% fat) for 14-17 weeks. We measured body composition, glucose and insulin tolerance, insulin response in peripheral tissues, energy expenditure, oxygen consumption, and respiratory exchange ratio in vivo. All these parameters were indistinguishable between wild type and knockout mice on chow diet and were modified equally by diet-induced obesity. Interestingly, we observed decreased food intake and ambulatory capacity of PUMA knockout mice on high fat diet. This was associated with increased adipocyte size and fasted leptin concentration in the blood. Our findings suggest that although PUMA is dispensable for glucose homeostasis in lean and obese mice, it can affect leptin levels and food intake during obesity.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/deficiencia , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Glucosa/metabolismo , Obesidad/fisiopatología , Proteínas Supresoras de Tumor/deficiencia , Tejido Adiposo/patología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Dieta Alta en Grasa/efectos adversos , Prueba de Tolerancia a la Glucosa , Homeostasis/fisiología , Insulina/farmacología , Resistencia a la Insulina , Leptina/sangre , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/patología , Proteínas Recombinantes/farmacología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
9.
Sci Rep ; 6: 21274, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26880502

RESUMEN

Aggregation of human islet amyloid polypeptide (hIAPP) into fibrils and plaques is associated with pancreatic ß-cell loss in type 2 diabetes (T2D). However, due to the rapidness of hIAPP conversion in aqueous phase, exactly which hIAPP species is responsible for the observed toxicity and through what mechanisms remains ambiguous. In light of the importance of understanding hIAPP toxicity for T2D here we show a biophysical scheme based on the use of a lipophilic Laurdan dye for examining MIN6 cell membranes upon exposure to fresh and oligomeric hIAPP as well as mature amyloid. It has been found that all three hIAPP species, especially fresh hIAPP, enhanced membrane fluidity and caused losses in cell viability. The cell generation of reactive oxygen species (ROS), however, was the most pronounced with mature amyloid hIAPP. The correlation between changes in membrane fluidity and cell viability and their lack of correlation with ROS production suggest hIAPP toxicity is elicited through both physical and biochemical means. This study offers a new insight into ß-cell toxicity induced by controlled hIAPP species, as well as new biophysical methodologies that may prove beneficial for the studies of T2D as well as neurological disorders.


Asunto(s)
Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Fluidez de la Membrana/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Especies Reactivas de Oxígeno/metabolismo
10.
Trends Endocrinol Metab ; 26(1): 30-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25432462

RESUMEN

Protein tyrosine phosphatases (PTPs) are a large family of enzymes that generally oppose the actions of protein tyrosine kinases (PTKs). Genetic polymorphisms for particular PTPs are associated with altered risk of both type 1 diabetes (T1D) and type 2 diabetes (T2D). Moreover, recent evidence suggests that PTPs play crucial roles in metabolism. They can act as regulators of liver homeostasis, food intake, or immune-mediated pancreatic b cell death. In this review we describe the mechanisms by which different members of the non-receptor PTP (PTPN) family influence metabolic physiology. This 'metabolic job' of PTPs is discussed in depth and the role of these proteins in different cell types compared. Understanding the pathways regulated by PTPs will provide novel therapeutic strategies for the treatment of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Genes de Cambio/fisiología , Metabolismo/genética , Proteínas Tirosina Fosfatasas/fisiología , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/genética , Humanos , Inmunidad Celular/genética , Células Secretoras de Insulina/metabolismo , Modelos Moleculares , Polimorfismo Genético , Proteínas Tirosina Fosfatasas/genética
11.
J Diabetes Res ; 2015: 280615, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26064977

RESUMEN

Pancreatic ß-cell loss induced by saturated free fatty acids (FFAs) is believed to contribute to type 2 diabetes. Previous studies have shown induction of endoplasmic reticulum (ER) stress, increased ubiquitinated proteins, and deregulation of the Bcl-2 family in the pancreas of type 2 diabetic patients. However, the precise mechanism of ß-cell death remains unknown. In the present study we demonstrate that the FFA palmitate blocks the ubiquitin-proteasome system (UPS) and causes apoptosis through induction of ER stress and deregulation of Bcl-2 proteins. We found that palmitate and the proteasome inhibitor MG132 induced ER stress in ß-cells, resulting in decreased expression of the prosurvival proteins Bcl-2, Mcl-1, and Bcl-XL, and upregulation of the prodeath BH3-only protein PUMA. On the other hand, pharmacological activation of the UPS by sulforaphane ameliorated ER stress, upregulated prosurvival Bcl-2 proteins, and protected ß-cells from FFA-induced cell death. Furthermore, transgenic overexpression of Bcl-2 protected islets from FFA-induced cell death in vitro and improved glucose-induced insulin secretion in vivo. Together our results suggest that targeting the UPS and Bcl-2 protein expression may be a valuable strategy to prevent ß-cell demise in type 2 diabetes.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ácido Palmítico/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ubiquitina/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Leupeptinas/farmacología , Ratones , Ubiquitinación/efectos de los fármacos
12.
Diabetes ; 64(7): 2489-96, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25732191

RESUMEN

Type 1 diabetes (T1D) is the result of an autoimmune assault against the insulin-producing pancreatic ß-cells, where chronic local inflammation (insulitis) leads to ß-cell destruction. T cells and macrophages infiltrate into islets early in T1D pathogenesis. These immune cells secrete cytokines that lead to the production of reactive oxygen species (ROS) and T-cell invasion and activation. Cytokine-signaling pathways are very tightly regulated by protein tyrosine phosphatases (PTPs) to prevent excessive activation. Here, we demonstrate that pancreata from NOD mice with islet infiltration have enhanced oxidation/inactivation of PTPs and STAT1 signaling compared with NOD mice that do not have insulitis. Inactivation of PTPs with sodium orthovanadate in human and rodent islets and ß-cells leads to increased activation of interferon signaling and chemokine production mediated by STAT1 phosphorylation. Furthermore, this exacerbated STAT1 activation-induced cell death in islets was prevented by overexpression of the suppressor of cytokine signaling-1 or inactivation of the BH3-only protein Bim. Together our data provide a mechanism by which PTP inactivation induces signaling in pancreatic islets that results in increased expression of inflammatory genes and exacerbated insulitis.


Asunto(s)
Interferón gamma/farmacología , Islotes Pancreáticos/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal/fisiología , Anciano , Animales , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT1/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda